US20130129840A1 - Combination therapy using a ruthenium complex - Google Patents

Combination therapy using a ruthenium complex Download PDF

Info

Publication number
US20130129840A1
US20130129840A1 US13/744,423 US201313744423A US2013129840A1 US 20130129840 A1 US20130129840 A1 US 20130129840A1 US 201313744423 A US201313744423 A US 201313744423A US 2013129840 A1 US2013129840 A1 US 2013129840A1
Authority
US
United States
Prior art keywords
cancer
tetrachlorobis
ruthenate
indazole
iii
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/744,423
Inventor
Hooshmand SHESHBARADARAN
Walter Berger
Petra Heffeter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Niiki Pharma Acquisition Corp 2
Original Assignee
Niiki Pharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Niiki Pharma Inc filed Critical Niiki Pharma Inc
Priority to US13/744,423 priority Critical patent/US20130129840A1/en
Publication of US20130129840A1 publication Critical patent/US20130129840A1/en
Assigned to NIIKI PHARMA ACQUISITION CORP. 2 reassignment NIIKI PHARMA ACQUISITION CORP. 2 ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NIIKI PHARMA INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/305Mercury compounds
    • A61K31/31Mercury compounds containing nitrogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention generally relates to method for treating cancer, and particularly to a method of treating cancer using trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof.
  • ruthenium complex compounds are known in the art to be useful as anti-tumor compounds. See e.g., U.S. Pat. No. 4,843,069, PCT Publication No. WO 9736595, and US Application Publication No. 2005032801.
  • the ruthenium complex salts indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (KP1099) and sodium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (KP1339) have been shown in preclinical studies to be effective in inducing apoptosis in colon cancer cells.
  • the present invention provides a method of treating cancer in a patient in need of such treatment comprising administering to the patient, simultaneously or sequentially, a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof and one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and
  • platinum agents e.g
  • the present invention further provides use of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for use in combination with one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib,
  • the present invention provides use of one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, rid
  • platinum agents
  • a kit comprising in a compartmentalized container a first unit dosage form having trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and a second unit dosage form of one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozo
  • platinum agents e
  • FIG. 1 shows a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the lung carcinoma cell line A549;
  • FIG. 2 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the colorectal carcinoma cell line HCT-116;
  • FIG. 3 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the gastric carcinoma cell line N87;
  • FIG. 4 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and oxaliplatin in the colorectal adenocarcinoma cell line LoVo;
  • FIG. 5 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and docetaxel in the prostate carcinoma cell line LNCap-1;
  • FIG. 6 shows an isobologram illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and docetaxelin the gastric carcinoma cell line N87.
  • Y axis is “Dose A” and X axis is “Dose B”;
  • FIG. 7 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the colorectal carcinoma cell line HCT-116;
  • FIG. 8 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the colorectal adenocarcinoma cell line LoVo;
  • FIG. 9 is a combination index plot illustrating the additive to synergistic activity between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the breast carcinoma cell line ZR-75-1;
  • FIG. 10 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and gemcitabine in the lung carcinoma cell line A549;
  • FIG. 11 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and gemcitabine in the pancreatic carcinoma cell line PANC-1;
  • FIG. 12 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in liver carcinoma cell line Hep3B2.1-7;
  • FIG. 13 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line A549;
  • FIG. 14 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and doxorubicinin the liver carcinoma cell line Hep 3B 2.1-7;
  • FIG. 15 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the lung carcinoma cell line A549;
  • FIG. 16 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density), E: erlotinib);
  • FIG. 17 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density), E: erlotinib);
  • FIG. 18 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and BCNU in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 19 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and BCNU in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 20 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sunitinib in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 21 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and temozolomide in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 22 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and temozolomide in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 23 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the hepatoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 24 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the hepatoma cell line HepG2 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 25 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line VL-8 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 26 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line A549 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 27 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the mesothelioma cell line P31 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density));
  • FIG. 28 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the melanoma cell line VM-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density), S: sorafenibs);
  • FIG. 29 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the colon cancer cell line SW480 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration ( ⁇ M), Y axis: O.D. (optical density), S: sorafenib);
  • FIG. 30 is a graph illustrating that the combination between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the Hep3B liver carcinoma xenograft model yields long term survival (Y-axis: % survival; X-axis: days);
  • FIG. 31 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and everolimus in the neuroendocrine tumor cell line MKL-1.
  • the present invention provides a method of treating cancer by a combination therapy.
  • the method comprises treating a cancer patient in need of treatment with a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, as well as one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel and paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin
  • the term “pharmaceutically acceptable salts” refers to the relatively non-toxic, organic or inorganic salts of the active compounds, including inorganic or organic salts of the compound.
  • Exemplary salts of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] include indazolium salt (e.g.,indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]), and alkali metal salts (e.g., sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]), etc.
  • the phrase “treating . . . with . . . ” means either administering a compound to the patient or causing the formation of a compound inside the patient.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) one or more anti-cancer agents chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel and paclitaxel),anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea
  • the method comprises administering a therapeutically effective amount of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) to a cancer patient who is under treatment of the one or more other anti-cancer agents provided above, or administering a therapeutically effective amount of such one or more other anti-cancer agents provided above to a cancer patient who is under treatment of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • a variety of cancers can be treated with the method of the present invention, including, but not limited to, brain cancer (e.g., astrocytoma such as glioblastoma), breast cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, lung cancer (NSCLC and small cell lung cancer), colorectal cancer, liver cancer (e.g., hepatocellular carcinoma), melanoma, pancreatic cancer, neuroendocrine tumors, prostate cancer, renal cancer, endometrial cancer, and sarcoma.
  • brain cancer e.g., astrocytoma such as glioblastoma
  • breast cancer ovarian cancer
  • cervical cancer gastric cancer
  • esophageal cancer esophageal cancer
  • lung cancer NSCLC and small cell lung cancer
  • colorectal cancer e.g., liver cancer (e.g., hepatocellular carcinoma), melanoma, pancreatic cancer, neuroendocrine tumors,
  • colorectal cancers such as colon carcinoma are treated with the combination method of the present invention.
  • liver cancers such as hepatocellular carcinoma are treated with the combination method of the present invention.
  • the combination therapy method of the present invention is used to treat melanoma.
  • lung cancer e.g. NSCLC and SCLC
  • gastroesophageal cancer e.g., gastric cancer, esophageal cancer
  • breast or ovarian cancer is treated with the combination therapy.
  • prostate cancer is treated with the combination therapy.
  • the combination therapy is applied to cervical or endometrial cancer.
  • kidney cancer is treated using the combination therapy method of the present invention.
  • the combination therapy is applied to pancreatic cancer.
  • the combination therapy is applied to neuroendocrine tumors.
  • a patient having cancer is identified or diagnosed, and the patient is treated with a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and a therapeutically effective amount of the one or more anti-cancer agents provided above.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) a platinum agent such as cisplatin, carboplatin and oxaliplatin.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • a platinum agent such as cisplatin, carboplatin and oxaliplatin.
  • the method is used for the treatment of colorectal cancer, lung cancer, or gastroesophageal cancer (e.g., gastric cancer or esophageal cancer).
  • the method is used for treating ovarian cancer, small cell lung cancer, testicular cancer, bladder carcinoma
  • the method is used for treating head and neck cancer, and brain tumors.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • oxaliplatin e.g., an alkali metal salt such as sodium salt
  • the combination of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) and (2) a platinum agent (e.g., cisplatin, carboplatin and oxaliplatin) is used for the treatment of lung cancer.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • a platinum agent e.g., cisplatin, carboplatin and oxaliplatin
  • the combination of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) and (2) a platinum agent (e.g., cisplatin, carboplatin and oxaliplatin) is used for the treatment of gastroesophageal cancer.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • a platinum agent e.g., cisplatin, carboplatin and oxaliplatin
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) a taxane (e.g., docetaxel, paclitaxel).
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • a taxane e.g., docetaxel, paclitaxel
  • the method is used for the treatment of prostate cancer, gastroesophageal cancer (e.g., gastric cancer) and lung cancer (e.g., non-small cell lung cancer).
  • the method comprises administering,simultaneously or sequentially, to a patient (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and (2) docetaxel for the treatment of breast cancer, lung cancer, prostate cancer, gastroesophageal cancer, or head and neck cancer.
  • the method comprises administering to a patient (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and (2) paclitaxel for the treatment of breast cancer, ovarian cancer, lung cancer, head and neck cancer, gastric cancer, esophagus cancer, bladder cancer, endometrial cancer, or cervical cancer.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) an anthracycline (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, particularly doxorubicin).
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • an anthracycline e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, particularly doxorubicin.
  • the combination comprising (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) doxorubicin is applied to treat liver cancer (e.g., hepatocellular carcinoma).
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • doxorubicin is applied to treat liver cancer (e.g., hepatocellular carcinoma).
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) 5-fluorouracil or a prodrug thereof (e.g., capecitabine, tegafur and S1).
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • 5-fluorouracil or a prodrug thereof e.g., capecitabine, tegafur and S1
  • the method is used for the treatment of colorectal cancer or breast cancer or pancreatic cancer.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) gemcitabine.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • gemcitabine e.g., an alkali metal salt such as sodium salt
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) an EGFR inhibitor.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • the method is applied to the treatment of lung cancer (e.g., NSCLC), pancreatic cancer, cervical cancer colorectal cancer, or liver cancer (particular hepatocellular carcinoma).
  • EGFR inhibitors are well known in the art, including, but not limited to, small molecule EGFR inhibitors (e.g., erlotinib, gefitinib, afatinib), and EGFR antibodies (cetuximab, panitumumab, nimotuzumab, necitumumab, etc.).
  • small molecule EGFR inhibitors e.g., erlotinib, gefitinib, afatinib
  • EGFR antibodies cetuximab, panitumumab, nimotuzumab, necitumumab, etc.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) sorafenib or regorafenib.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • sorafenib or regorafenib e.g., an alkali metal salt such as sodium salt
  • the cancer treated is liver cancer (e.g., hepatocellular carcinoma), lung cancer (e.g., NSCLC), colorectal cancer or melanoma.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) sunitinib.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • sunitinib e.g., an alkali metal salt such as sodium salt
  • the combination is used to treat liver cancer (e.g., hepatocellular carcinoma).
  • the combination is used to treat renal cell carcinoma, gastrointestinal stromal tumor, and neuroendocrine tumors.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) temozolomide.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • the combination is used for the treatment of liver cancer, brain cancer (e.g., glioblastoma) or melanoma.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) BCNU.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • the combination therapy is used for the treatment of liver cancer or cervical cancer or brain cancer.
  • the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) one or more mTOR inhibitors.
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • mTOR inhibitors include, but not limited to, e.g., everolimus, temsirolimus, ridaforolimus, sirolimus etc.
  • the combination therapy is used for the treatment of neuroendocrine tumors (NET), kidney cancer, astrocytoma, breast cancer, gastric cancer, or hepatocellular carcinoma.
  • the combination therapy comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) everolimus for treating neuroendocrine tumors (NET).
  • a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] e.g., an alkali metal salt such as sodium salt
  • NET neuroendocrine tumors
  • Alkali metal salts of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] are known in the art and disclosed in e.g., European Patent No. EP 0835112 B1, and can be made in any methods known in the art.
  • PCT Publication No. WO/2008/154553 discloses an efficient method of making sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
  • Indazolium salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] is disclosed in U.S. Pat. No. 7,338,946.
  • trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof and the one or more other anti-cancer agents can be administered at about the same time, or separately according to their respective dosing schedules.
  • the trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof can be administered in the same pharmaceutical composition or in separate dosage unit forms.
  • Trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and pharmaceutically acceptable salts thereof, such as sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be administered through intravenous injection or any other suitable means at a dosing of from 0.1 mg to 1000 mg per kg of body weight of the patient based on total body weight.
  • the active ingredients may be administered at once, or may be divided into a number of smaller doses to be administered at predetermined intervals of time, e.g., once daily or once every two days. See e.g., Hartinger et al., J. Inorg. Biochem., 100:891-904 (2006).
  • Injectable forms are generally known in the art, e.g., in buffered solution or suspension.
  • the other anti-cancer agents used in combination with a salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be administered through a route and at an amount generally recommended by their manufacturers or known in the art, e.g., as provided in the prescribing information sheet or product package insert as approved by relevant regulatory authorities, or varied therefrom, e.g., by one order of magnitude as clinicians see fit to accommodate specific patient situations.
  • the therapeutically effective amount for each active compound can vary with factors including but not limited to the activity of the compound used, stability of the active compound in the patient's body, the severity of the conditions to be alleviated, the total weight of the patient treated, the route of administration, the ease of absorption, distribution, and excretion of the active compound by the body, the age and sensitivity of the patient to be treated, and the like, as will be apparent to a skilled artisan.
  • the amount of administration can be adjusted as the various factors change over time.
  • a pharmaceutical kit comprising in a compartmentalized container (1) a unit dosage form of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate (III)], such as sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]; and (2) a unit dosage form of at least one (one, two, or more) anti-cancer agent chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof
  • platinum agents e.g., c
  • the amount of a therapeutic compound in the unit dosage form is determined by the dosage to be used on a patient in the method of the present invention.
  • a pharmaceutically acceptable salt trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be in lyophilized form in an amount of, e.g., 25 mg, in an ampoule.
  • the other anti-cancer agents to be used in the combination therapy and included in the kit can be in any dosage form generally known or used in the art, e.g., tablet, capsule, a lyophilized form for reconstitution of an injectable form, etc.
  • the kit further comprises instructions for using the kit in the combination therapy method in accordance with the present invention.
  • Human tumor cell lines including A549, HCT-116, Hep 3B2.1-7, LNCap clone FGC, LoVo, N87, PANC-1 and ZR-75-1 were obtained from the American Type Culture Collection (ATCC) or the UNC Lineberger Comprehensive Cancer Center.
  • the MKL-1 human neuroendocrine skin carcinoma cell line was obtained from the ECACC (European Collection of Cell Cultures).
  • Cell cultures were established using standard in vitro culture methods and supplier recommended media and supplements in 175 cm 2 Greiner® or Corning® tissue culture-treated flasks. All cell cultures were incubated in a humidified 37° C., 5% CO 2 , 95% air environment. The cells were sub-cultured regularly to maintain log phase growth.
  • the cells for each line were processed and seeded into 96-well cell culture-treated plates one cell line at a time.
  • the cells were removed from their culture flasks using trypsin solution pooled in a sterile conical tube and centrifuged at 350 ⁇ g for 5 minutes at room temperature. For MKL-1 cells in suspension, the cells did not require trypsinization.
  • the cell suspensions were diluted (based on live cell counts) using complete media to yield a final suspension density (cells/ml) based on previously determined seeding densities for each cell line for a 72 hour 96-well plate assay.
  • the tissue culture treated plates for EC 50 testing were seeded at a density specified below in Table 1 and incubated overnight at 37° C. in a 5% CO 2 , 95% air humidified atmosphere to allow the cells to attach.
  • Test Agent Preparation For each single agent or combination of test agents, the top concentration mixture (2 ⁇ final treatment concentration) was made in sterile 1.5 ml microcentrifuge tubes and then directly transferred to the first well of the treatment dilution plates. A 200 mM stock solution of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (“test drug”) was made using 500 ⁇ l of 100% dimethyl sulfoxide (DMSO). An aliquot of the 200 mM stock solution was used to also make a 40 mM stock solution in 100% DMSO (10 ⁇ l of 200 mM stock+40 ⁇ l DMSO for the N87 cell line).
  • DMSO dimethyl sulfoxide
  • 5-Fluororuracil was manufactured by TEVA Parenteral Medicines and supplied in vials at a concentration of 50 mg/ml (384.4 mM) in aqueous solution.
  • Cisplatin was obtained from Sigma-Aldrich, and a 4 mM stock solution of cisplatin was made using 0.9% saline and stored at ⁇ 20° C. After thawing, the 4 mM stock solution was diluted 2 ⁇ using complete media to yield a 2 mM solution in the first well of a 96-well dilution plate for the positive control test plate wells. This was then serially diluted 1:4 in complete media across nine wells for a total of ten concentrations ranging from 2,000-0.008 ⁇ M. The 4 mM stock solution was also diluted for use as a single standard agent and in combination with sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
  • Docetaxel manufactured by Fluka was weighed out (1.6 mg) and a 2,000 ⁇ M solution was made by adding 0.990 ml 100% DMSO and intermittently vortexing for 1-15 seconds. This was further diluted in 100% DMSO to make a 40 ⁇ M stock solution (10 ⁇ l of 2,000 ⁇ M docetaxel+490 ⁇ l DMSO).
  • Gemcitabine manufactured by Eli Lilly and Company
  • a 50 mM clear, colorless stock solution was made by adding 188 ⁇ l of sterile water. This was further diluted 1,000 ⁇ in complete media to yield a 50 ⁇ M stock solution (10 ⁇ l of 50 mM gemcitabine+9.990 ml media).
  • Sorafenib was obtained from LC Laboratories and a 100 mM stock solution was made by adding 0.188 ml of 100% DMSO to 12.0 mg sorafenib.
  • Everolimus was obtained from LC Laboratories and a 48 mM clear, colorless stock solution was made by adding 117 ⁇ l of 100% DMSO to 5.4 mg of everolimus.
  • the antiproliferative activity of the test agents was evaluated using the MTT Cell Proliferation Assay Kit (ATCC catalog #30-1010K).
  • the MTT assay is based on the reduction of yellow tetrazolium MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) by metabolically active cells forming purple formazan crystals.
  • the purple formazan is solubilized with detergent and quantified spectrophotometrically at 570 nm.
  • Test agents were serially diluted in complete culture media (+1% DMSO where appropriate) and added to each well in a volume of 0.1 mL for a total final volume of 0.2 mL/well (0.5% DMSO final, where used). Cells were exposed to test agents for 72 hours.
  • Absorbance values were converted to Percent of Control and plotted against test agent concentrations for EC 50 calculations using SoftMax® Pro (version 5.2, Molecular Devices). The plate blank signal average was subtracted from all wells prior to calculating the Percent of Control. Percent of Control values were calculated by dividing the absorbance values for each test well by the No Drug Control average (column 11 values; cells+vehicle control) and multiplying by 100. Plots of Compound Concentration vs. Percent of Control were analyzed using the 4-parameter equation to obtain EC 50 values and other parameters that describe the sigmoidal dose response curve.
  • Combination data was analyzed using CompuSyn® software to calculate Combination Index (CI) values to assess synergy.
  • the Fractional Affect (Fa) was calculated from the Percent of Control (from SoftMax® Pro) using the formula: 1 ⁇ (Percent Control/100).
  • the dosage, fractional affect and molar ratio of compounds tested in combination were entered into the CompuSyn® software for evaluation of the presence/absence of synergy.
  • CompuSyn® assigns a Combination Index (CI) value which rates the level of compounds' effect on proliferation.
  • CI values below 1 indicate the presence of synergy and CI values above 1 indicate antagonism.
  • CI values close to 1 indicate an additive affect. See Chou, P HARMACOL. R EV., 58(3):621-81(2006).
  • Table 2 summarizes the CI values of the synergistic combinations.
  • the hepatocellular carcinoma cell line Hep3B (from ATCC) was grown in RMPI 1640 supplemented with 10% fetal bovine serum.
  • the epidermal carcinoma-derived cell line KB-3-1 was grown in RPMI 1640+10% FCS. See Shen et al., J. Biol. Chem., 261:7762-7770 (1986).
  • Cytotoxicity Assays Cells were plated (2 ⁇ 10 3 cells in 100 ⁇ l/well) in 96-well plates and allowed to recover for 24 hours. Drugs were added in another 100 ⁇ l growth medium and cells exposed for 72 hours. The proportion of viable cells was determined by MTT assay following the manufacturer's recommendations (EZ4U, Biomedica, Vienna, Austria).
  • FIGS. 16-22 significant synergies were exhibited by the combination of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and anti-cancer drugs including erlotinib ( FIGS. 16 and 17 ), BCNU ( FIGS. 18 and 19 ), sunitinib ( FIG. 20 ), and temozolomide ( FIGS. 21 and 22 ) in the cell lines tested.
  • Sorafenib was purchased from LC Laboratories (Woburn, USA). All other substances were purchased from Sigma-Aldrich (St. Louis, USA).
  • the hepatocellular carcinoma cell line Hep3B was purchased from American Type Culture Collection, Manassas, Va. Cells were grown in RMPI 1640 supplemented with 10% fetal bovine serum. The colon carcinoma cell line HCT116 and respective subline with deleted p53 genes were grown in McCoy's culture medium supplemented with 10% FCS. See Bunz et al, Cancer Res., 62:1129-1133 (2002). Lung cancer cell line A549 was grown in RPMI 1640 medium with 10% FCS, and the hepatocellular carcinoma cell line HepG2 was cultured in the Minimal Essential Medium supplemented with non-essential aminoacids, pyruvate, and 10% FCS.
  • Cytotoxicity Assays Cells were plated (2 ⁇ 10 3 cells in 100 ⁇ l/well) in 96-well plates and allowed to recover for 24 hours. Drugs were added in another 100 ⁇ l growth medium and cells exposed for 72 hours. The proportion of viable cells was determined by MTT assay following the manufacturer's recommendations (EZ4U, Biomedica, Vienna, Austria).
  • sorafenib and sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] gives rise to significant synergies in a variety of cell lines including hepatocellular carcinoma cell line Hep3B, hepatocellular carcinoma cell line HepG2, lung carcinoma cell line VL-8, lung carcinoma cell line A549, mesothelioma cell line P31, colon cancer cell line SW480, and melanoma cell line VM-1.
  • CB17 severe combined immunodeficient (SCID) female mice were used for all in vivo studies. The mice received food and water ad libitum. For tumor application, logarithmically growing Hep3B cells in cell culture were collected by trypsinization and washed once in serum-free culture medium. The cells were then pelleted and resuspended in culture medium to a final cell count of 2 ⁇ 10 7 /ml. 50 ⁇ l of the cell suspension was injected s.c. in the right flank of each mouse. Treatment started when all animals in the study had established tumors of a size of about 3 ⁇ 3 mm.
  • SCID severe combined immunodeficient
  • Tumor size was calculated using the equation (l ⁇ w 2 )/2, where l and w refer to the larger and smaller dimensions, respectively, of the tumor. 4 mice were used in each group for each data point.
  • the purpose of this experiment was to evaluate the efficacy of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)], administered intravenously (IV) as a single agent and in combination with cisplatin against early stage N87 human gastric carcinoma xenografts in female nude mice.
  • mice Female athymic mice (Hsd: Athymic Nude-Foxn1nu) were obtained from Harlan. They were 8 weeks old on Day 1 of the experiment. The mice were fed irradiated Rodent Diet 5053 (LabDietTM) and water ad libitum, and grown and experimented on in a clean and controlled environment. Test mice were implanted subcutaneously on Day 0 with 30 to 60 mg tumor fragments. All mice were observed for clinical signs at least once daily. Mice with tumors in excess of 1 g or with ulcerated tumors were euthanized. All procedures carried out in this experiment were conducted in compliance with all the laws, regulations and guidelines of the National Institutes of Health (NIH) and with the necessary approvals.
  • NASH National Institutes of Health
  • mice were weighed ⁇ 18.2 g at the initiation of therapy. Mean group body weights at first treatment were well-matched (range of group means, 22.3-22.8 g). All mice were dosed according to individual body weight on the day of treatment (0.2 ml/20 g). Sixteen days after the initial course of treatment was completed, a second course of treatment was begun for the combination groups only (groups in which cisplatin was dosed at 7.5 mg/kg). A complete second course of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] was given, but only two of the three planned doses of cisplatin were completed due to extensive weight loss.
  • the primary endpoints used to evaluate efficacy were: % T/C, tumor growth delay, and the number of tumor-free survivors (TFS) at the end of the study.
  • % T/C is defined as the median tumor mass of the Treated Group divided by the median tumor mass of the Control Group ⁇ 100. In this experiment, % T/C was evaluated when the median Control reached 1 g.
  • Tumor Growth Delay (T ⁇ C) was also used to quantify efficacy. Tumor growth delay for this experiment was expressed as a T ⁇ C value, where T and C are the median times in days required for the treatment and control group tumors, respectively, to grow to a selected evaluation size, 750 mg.
  • the purpose of this experiment was to evaluate the efficacy of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] as a single agent and in combination with paclitaxel against early stage A549 human lung carcinoma xenografts in female nude mice.
  • Sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] was administered intravenously every two days for three treatments and paclitaxel was administered intravenously for five consecutive days, both beginning on Day 3 post implant.
  • the animals were grown, implanted with tumors and experimented on in the same manner as in Example 5 above, unless otherwise clarified below.
  • Cremophor EL® was used in the context of paclitaxel administration. Specifically, on each day of treatment, the paclitaxel was dissolved in absolute ethanol (10% of the final volume), followed by sequential addition of Cremophor EL® (10% of the final volume) and saline (80% of the final volume) with thorough mixing after each addition.
  • mice were weighed ⁇ 17.3 g at the initiation of therapy. Mean group body weights at first treatment were well-matched (range of group means, 20.6-23.5 g). All mice were dosed according to individual body weight on the day of treatment (0.2 ml/20 g).

Abstract

A combination therapy is disclosed for treating cancer. The method comprises administering to a cancer patient a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and administering to the patient a therapeutically effective amount of one or more other anti-cancer agents as disclosed herein.

Description

    RELATED APPLICATIONS
  • This is a continuation of PCT/US11/44302 filed on Jul. 18, 2011, which claims the priority of U.S. Provisional Application No. 61/365,329 filed on Jul. 18, 2010, the entire content of both of which being incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention generally relates to method for treating cancer, and particularly to a method of treating cancer using trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof.
  • BACKGROUND OF THE INVENTION
  • A number of ruthenium complex compounds are known in the art to be useful as anti-tumor compounds. See e.g., U.S. Pat. No. 4,843,069, PCT Publication No. WO 9736595, and US Application Publication No. 2005032801. In particular, the ruthenium complex salts indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (KP1099) and sodium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (KP1339) have been shown in preclinical studies to be effective in inducing apoptosis in colon cancer cells. See e.g., Kapitza et al., J. Cancer Res. Clin. Oncol., 131(2):101-10 (2005). In addition, the compound ruthenium complex salt indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (KP1019) showed some anti-cancer activities in a phase I clinical trial.
  • SUMMARY OF THE INVENTION
  • It has been discovered that the combined use of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and a number of other anti-cancer compounds creates significant synergies in the treatment of cancers. Accordingly, in a first aspect, the present invention provides a method of treating cancer in a patient in need of such treatment comprising administering to the patient, simultaneously or sequentially, a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof and one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus, etc.), sorafenib, regorafenib, and sunitinib.
  • The present invention further provides use of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for use in combination with one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus, etc.), sorafenib, regorafenib, and sunitinib, for treating, preventing or delaying the onset of cancer.
  • To put it differently, the present invention provides use of one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus, etc.), sorafenib, regorafenib, and sunitinib, for the manufacture of a medicament for use in combination with trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof in treating, preventing or delaying the onset of cancer.
  • In yet another aspect, a kit is provided comprising in a compartmentalized container a first unit dosage form having trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and a second unit dosage form of one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab etc.), mTOR inhibitors (e.g., everolimus, temsirolimus,ridaforolimus, sirolimus etc.), sorafenib, regorafenib, and sunitinib. Optionally, instructions on how to use the kit are included in the kit.
  • The foregoing and other advantages and features of the invention, and the manner in which the same are accomplished, will become more readily apparent upon consideration of the following detailed description of the invention taken in conjunction with the accompanying examples, which illustrate preferred and exemplary embodiments.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the lung carcinoma cell line A549;
  • FIG. 2 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the colorectal carcinoma cell line HCT-116;
  • FIG. 3 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and cisplatin in the gastric carcinoma cell line N87;
  • FIG. 4 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and oxaliplatin in the colorectal adenocarcinoma cell line LoVo;
  • FIG. 5 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and docetaxel in the prostate carcinoma cell line LNCap-1;
  • FIG. 6 shows an isobologram illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and docetaxelin the gastric carcinoma cell line N87.Y axis is “Dose A” and X axis is “Dose B”;
  • FIG. 7 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the colorectal carcinoma cell line HCT-116;
  • FIG. 8 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the colorectal adenocarcinoma cell line LoVo;
  • FIG. 9 is a combination index plot illustrating the additive to synergistic activity between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and 5-FU in the breast carcinoma cell line ZR-75-1;
  • FIG. 10 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and gemcitabine in the lung carcinoma cell line A549;
  • FIG. 11 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and gemcitabine in the pancreatic carcinoma cell line PANC-1;
  • FIG. 12 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in liver carcinoma cell line Hep3B2.1-7;
  • FIG. 13 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line A549;
  • FIG. 14 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and doxorubicinin the liver carcinoma cell line Hep 3B 2.1-7;
  • FIG. 15 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the lung carcinoma cell line A549;
  • FIG. 16 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density), E: erlotinib);
  • FIG. 17 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and erlotinib in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density), E: erlotinib);
  • FIG. 18 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and BCNU in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 19 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and BCNU in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 20 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sunitinib in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 21 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and temozolomide in the liver carcinoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 22 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and temozolomide in the cervix carcinoma cell line KB-3-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 23 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the hepatoma cell line Hep3B (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 24 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the hepatoma cell line HepG2 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 25 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line VL-8 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 26 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the lung carcinoma cell line A549 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 27 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the mesothelioma cell line P31 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density));
  • FIG. 28 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the melanoma cell line VM-1 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density), S: sorafenibs);
  • FIG. 29 is a graph showing the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the colon cancer cell line SW480 (X axis: sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] concentration (μM), Y axis: O.D. (optical density), S: sorafenib);
  • FIG. 30 is a graph illustrating that the combination between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and sorafenib in the Hep3B liver carcinoma xenograft model yields long term survival (Y-axis: % survival; X-axis: days);
  • FIG. 31 is a combination index plot illustrating the synergism between sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and everolimus in the neuroendocrine tumor cell line MKL-1.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a method of treating cancer by a combination therapy. The method comprises treating a cancer patient in need of treatment with a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, as well as one or more drugs chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel and paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus, etc.), sorafenib, regorafenib, and sunitinib. As used herein, the term “pharmaceutically acceptable salts” refers to the relatively non-toxic, organic or inorganic salts of the active compounds, including inorganic or organic salts of the compound. Exemplary salts of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] include indazolium salt (e.g.,indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]), and alkali metal salts (e.g., sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]), etc. As used herein, the phrase “treating . . . with . . . ” means either administering a compound to the patient or causing the formation of a compound inside the patient.
  • In some embodiments, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) one or more anti-cancer agents chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel and paclitaxel),anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and S1), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus, etc.), sorafenib, regorafenib, and sunitinib. To put it differently, in accordance with this embodiment, the method comprises administering a therapeutically effective amount of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) to a cancer patient who is under treatment of the one or more other anti-cancer agents provided above, or administering a therapeutically effective amount of such one or more other anti-cancer agents provided above to a cancer patient who is under treatment of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
  • A variety of cancers can be treated with the method of the present invention, including, but not limited to, brain cancer (e.g., astrocytoma such as glioblastoma), breast cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, lung cancer (NSCLC and small cell lung cancer), colorectal cancer, liver cancer (e.g., hepatocellular carcinoma), melanoma, pancreatic cancer, neuroendocrine tumors, prostate cancer, renal cancer, endometrial cancer, and sarcoma.
  • In one embodiment, colorectal cancers such as colon carcinoma are treated with the combination method of the present invention. In another embodiment, liver cancers such as hepatocellular carcinoma are treated with the combination method of the present invention. In another embodiment, the combination therapy method of the present invention is used to treat melanoma. In another embodiment, lung cancer (e.g. NSCLC and SCLC) is treated with the combination therapy method. In yet another embodiment, gastroesophageal cancer (e.g., gastric cancer, esophageal cancer) is treated with the combination therapy. In another embodiment, breast or ovarian cancer is treated with the combination therapy. In yet another embodiment, prostate cancer is treated with the combination therapy. In yet another embodiment, the combination therapy is applied to cervical or endometrial cancer. In yet another embodiment, kidney cancer is treated using the combination therapy method of the present invention. In yet another embodiment, the combination therapy is applied to pancreatic cancer. In another embodiment, the combination therapy is applied to neuroendocrine tumors.
  • Thus, in these various embodiments, in accordance with the present invention, a patient having cancer is identified or diagnosed, and the patient is treated with a therapeutically effective amount of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof, and a therapeutically effective amount of the one or more anti-cancer agents provided above.
  • In one embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) a platinum agent such as cisplatin, carboplatin and oxaliplatin. In specific embodiments, the method is used for the treatment of colorectal cancer, lung cancer, or gastroesophageal cancer (e.g., gastric cancer or esophageal cancer). In other specific embodiments, the method is used for treating ovarian cancer, small cell lung cancer, testicular cancer, bladder carcinoma. In other specific embodiments, the method is used for treating head and neck cancer, and brain tumors. In one specific embodiment, the combination of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) and (2) oxaliplatin is used for the treatment of colorectal cancer. In one specific embodiment, the combination of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) and (2) a platinum agent (e.g., cisplatin, carboplatin and oxaliplatin) is used for the treatment of lung cancer. In another specific embodiment, the combination of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt) and (2) a platinum agent (e.g., cisplatin, carboplatin and oxaliplatin) is used for the treatment of gastroesophageal cancer.
  • In another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) a taxane (e.g., docetaxel, paclitaxel). In some specific embodiments, the method is used for the treatment of prostate cancer, gastroesophageal cancer (e.g., gastric cancer) and lung cancer (e.g., non-small cell lung cancer). In some specific embodiments, the method comprises administering,simultaneously or sequentially, to a patient (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and (2) docetaxel for the treatment of breast cancer, lung cancer, prostate cancer, gastroesophageal cancer, or head and neck cancer. In some other specific embodiments, the method comprises administering to a patient (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and (2) paclitaxel for the treatment of breast cancer, ovarian cancer, lung cancer, head and neck cancer, gastric cancer, esophagus cancer, bladder cancer, endometrial cancer, or cervical cancer.
  • In another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) an anthracycline (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, particularly doxorubicin). In some specific embodiments, the combination comprising (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) doxorubicin is applied to treat liver cancer (e.g., hepatocellular carcinoma).
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) 5-fluorouracil or a prodrug thereof (e.g., capecitabine, tegafur and S1). In specific embodiments, the method is used for the treatment of colorectal cancer or breast cancer or pancreatic cancer.
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) gemcitabine. In some specific embodiments, the cancer treated is pancreatic cancer, lung cancer, bladder cancer or breast cancer.
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) an EGFR inhibitor. In some specific embodiments, the method is applied to the treatment of lung cancer (e.g., NSCLC), pancreatic cancer, cervical cancer colorectal cancer, or liver cancer (particular hepatocellular carcinoma). EGFR inhibitors are well known in the art, including, but not limited to, small molecule EGFR inhibitors (e.g., erlotinib, gefitinib, afatinib), and EGFR antibodies (cetuximab, panitumumab, nimotuzumab, necitumumab, etc.).
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) sorafenib or regorafenib. In specific embodiments, the cancer treated is liver cancer (e.g., hepatocellular carcinoma), lung cancer (e.g., NSCLC), colorectal cancer or melanoma.
  • In another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) sunitinib. In specific embodiments, the combination is used to treat liver cancer (e.g., hepatocellular carcinoma). In other specific embodiments, the combination is used to treat renal cell carcinoma, gastrointestinal stromal tumor, and neuroendocrine tumors.
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) temozolomide. In specific embodiments, the combination is used for the treatment of liver cancer, brain cancer (e.g., glioblastoma) or melanoma.
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) BCNU. In some specific embodiments, the combination therapy is used for the treatment of liver cancer or cervical cancer or brain cancer.
  • In yet another embodiment, the method of treating cancer comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) one or more mTOR inhibitors. Examples of mTOR inhibitors include, but not limited to, e.g., everolimus, temsirolimus, ridaforolimus, sirolimus etc. In some specific embodiments, the combination therapy is used for the treatment of neuroendocrine tumors (NET), kidney cancer, astrocytoma, breast cancer, gastric cancer, or hepatocellular carcinoma. In some specific embodiments, the combination therapy comprises administering to a cancer patient in need of treatment, simultaneously or sequentially, a therapeutically effective amount of (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (e.g., an alkali metal salt such as sodium salt), and (2) everolimus for treating neuroendocrine tumors (NET).
  • Alkali metal salts of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] are known in the art and disclosed in e.g., European Patent No. EP 0835112 B1, and can be made in any methods known in the art. For example, PCT Publication No. WO/2008/154553 discloses an efficient method of making sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]. Indazolium salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] is disclosed in U.S. Pat. No. 7,338,946.
  • The trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof and the one or more other anti-cancer agents can be administered at about the same time, or separately according to their respective dosing schedules. When administered at about the same time, the trans-[tetrachlorobis(1H-indazole)ruthenate(III)] or a pharmaceutically acceptable salt thereof can be administered in the same pharmaceutical composition or in separate dosage unit forms. Trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and pharmaceutically acceptable salts thereof, such as sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be administered through intravenous injection or any other suitable means at a dosing of from 0.1 mg to 1000 mg per kg of body weight of the patient based on total body weight. The active ingredients may be administered at once, or may be divided into a number of smaller doses to be administered at predetermined intervals of time, e.g., once daily or once every two days. See e.g., Hartinger et al., J. Inorg. Biochem., 100:891-904 (2006). Injectable forms are generally known in the art, e.g., in buffered solution or suspension.
  • The other anti-cancer agents used in combination with a salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be administered through a route and at an amount generally recommended by their manufacturers or known in the art, e.g., as provided in the prescribing information sheet or product package insert as approved by relevant regulatory authorities, or varied therefrom, e.g., by one order of magnitude as clinicians see fit to accommodate specific patient situations.
  • It should be understood that the dosage ranges set forth above are exemplary only and are not intended to limit the scope of this invention. The therapeutically effective amount for each active compound can vary with factors including but not limited to the activity of the compound used, stability of the active compound in the patient's body, the severity of the conditions to be alleviated, the total weight of the patient treated, the route of administration, the ease of absorption, distribution, and excretion of the active compound by the body, the age and sensitivity of the patient to be treated, and the like, as will be apparent to a skilled artisan. The amount of administration can be adjusted as the various factors change over time.
  • In accordance with another aspect of the present invention, a pharmaceutical kit is provided comprising in a compartmentalized container (1) a unit dosage form of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate (III)], such as sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]; and (2) a unit dosage form of at least one (one, two, or more) anti-cancer agent chosen from the group consisting of platinum agents (e.g., cisplatin, carboplatin, oxaliplatin, and picoplatin), taxane (e.g., docetaxel, paclitaxel), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), 5-FU and prodrugs thereof (e.g., capecitabine, tegafur and Si), nitrosourea compounds (e.g., carmustine (BCNU), lomustine (CCNU), semustine, ethylnitrosourea (ENU) and streptozotocin), gemcitabine, temozolomide, EGFR inhibitors (e.g., erlotinib, gefitinib, cetuximab, panumutimab), mTOR inhibitors (e.g., everolimus, temsirolimus, ridaforolimus, sirolimus etc.), sorafenib, regorafenib, and sunitinib. As will be apparent to a skilled artisan, the amount of a therapeutic compound in the unit dosage form is determined by the dosage to be used on a patient in the method of the present invention. In the kit, a pharmaceutically acceptable salt trans-[tetrachlorobis(1H-indazole)ruthenate(III)] can be in lyophilized form in an amount of, e.g., 25 mg, in an ampoule. The other anti-cancer agents to be used in the combination therapy and included in the kit can be in any dosage form generally known or used in the art, e.g., tablet, capsule, a lyophilized form for reconstitution of an injectable form, etc. Optionally, the kit further comprises instructions for using the kit in the combination therapy method in accordance with the present invention.
  • EXAMPLE 1
  • Cell Culture: Human tumor cell lines including A549, HCT-116, Hep 3B2.1-7, LNCap clone FGC, LoVo, N87, PANC-1 and ZR-75-1 were obtained from the American Type Culture Collection (ATCC) or the UNC Lineberger Comprehensive Cancer Center. The MKL-1 human neuroendocrine skin carcinoma cell line was obtained from the ECACC (European Collection of Cell Cultures). Cell cultures were established using standard in vitro culture methods and supplier recommended media and supplements in 175 cm2 Greiner® or Corning® tissue culture-treated flasks. All cell cultures were incubated in a humidified 37° C., 5% CO2, 95% air environment. The cells were sub-cultured regularly to maintain log phase growth.
  • On the day of EC50 plate seeding, the cells for each line were processed and seeded into 96-well cell culture-treated plates one cell line at a time. The cells were removed from their culture flasks using trypsin solution pooled in a sterile conical tube and centrifuged at 350×g for 5 minutes at room temperature. For MKL-1 cells in suspension, the cells did not require trypsinization.
  • The cell suspensions were diluted (based on live cell counts) using complete media to yield a final suspension density (cells/ml) based on previously determined seeding densities for each cell line for a 72 hour 96-well plate assay. The tissue culture treated plates for EC50 testing were seeded at a density specified below in Table 1 and incubated overnight at 37° C. in a 5% CO2, 95% air humidified atmosphere to allow the cells to attach.
  • TABLE 1
    Seeding Density for EC50 Assay
    Cells/well
    Cell Line Type (×103)
    A549 lung cancer 2.5
    HCT-116 colorectal cancer 8.0
    Hep 3B2.1-7 hepatocellular 6.0
    carcinoma
    LNCaP prostate cancer 4.0
    LoVo colorectal cancer 12.0
    N87 gastric cancer 20.0
    PANC-1 pancreatic cancer 6.0
    ZR-75-1 breast cancer 3.0
    MKL-1 human neuroendocrine 34
    skin carcinoma
  • Test Agent Preparation: For each single agent or combination of test agents, the top concentration mixture (2× final treatment concentration) was made in sterile 1.5 ml microcentrifuge tubes and then directly transferred to the first well of the treatment dilution plates. A 200 mM stock solution of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (“test drug”) was made using 500 μl of 100% dimethyl sulfoxide (DMSO). An aliquot of the 200 mM stock solution was used to also make a 40 mM stock solution in 100% DMSO (10 μl of 200 mM stock+40 μl DMSO for the N87 cell line).
  • 5-Fluororuracil was manufactured by TEVA Parenteral Medicines and supplied in vials at a concentration of 50 mg/ml (384.4 mM) in aqueous solution.
  • Cisplatin was obtained from Sigma-Aldrich, and a 4 mM stock solution of cisplatin was made using 0.9% saline and stored at −20° C. After thawing, the 4 mM stock solution was diluted 2× using complete media to yield a 2 mM solution in the first well of a 96-well dilution plate for the positive control test plate wells. This was then serially diluted 1:4 in complete media across nine wells for a total of ten concentrations ranging from 2,000-0.008 μM. The 4 mM stock solution was also diluted for use as a single standard agent and in combination with sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
  • Docetaxel manufactured by Fluka was weighed out (1.6 mg) and a 2,000 μM solution was made by adding 0.990 ml 100% DMSO and intermittently vortexing for 1-15 seconds. This was further diluted in 100% DMSO to make a 40 μM stock solution (10 μl of 2,000 μM docetaxel+490 μl DMSO).
  • 8.2 mg of Erlotinib (from LC Laboratories) was weighed out and a 50 mM cloudy, white suspension was made by adding 0.382 ml 100% DMSO and intermittently vortexing for 15-30 seconds.
  • 5.8 mg of Gemcitabine (manufactured by Eli Lilly and Company) was weighed out and a 50 mM clear, colorless stock solution was made by adding 188 μl of sterile water. This was further diluted 1,000× in complete media to yield a 50 μM stock solution (10 μl of 50 mM gemcitabine+9.990 ml media).
  • Sorafenib was obtained from LC Laboratories and a 100 mM stock solution was made by adding 0.188 ml of 100% DMSO to 12.0 mg sorafenib.
  • Everolimus was obtained from LC Laboratories and a 48 mM clear, colorless stock solution was made by adding 117 μl of 100% DMSO to 5.4 mg of everolimus.
  • EC50 Assay: The antiproliferative activity of the test agents was evaluated using the MTT Cell Proliferation Assay Kit (ATCC catalog #30-1010K). The MTT assay is based on the reduction of yellow tetrazolium MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) by metabolically active cells forming purple formazan crystals. The purple formazan is solubilized with detergent and quantified spectrophotometrically at 570 nm.
  • Cells in the log phase of growth were seeded at the indicated densities listed in Table 1 above into 96-well culture treated plates in 0.1 mL of complete media in all wells except for one column reserved for the media only control. The cells (except for the MKL-1 cells) were allowed to attach during an overnight incubation prior to treating with test agents. Test agents were serially diluted in complete culture media (+1% DMSO where appropriate) and added to each well in a volume of 0.1 mL for a total final volume of 0.2 mL/well (0.5% DMSO final, where used). Cells were exposed to test agents for 72 hours. Following the exposure to test agents, 0.1 mL of culture supernatant was carefully removed from all wells of each plate and 0.01 mL of MTT reagent was added to each well. The plates were returned to the incubator for four hours. Following the incubation period, kit supplied detergent reagent (0.1 mL) was added to all wells. The plates were wrapped in plastic wrap to prevent evaporation and allowed to sit at room temperature in the dark overnight. The absorbance at 570 nm was measured the following day using a SpectraMAX Plus plate reader (Molecular Devices).
  • Absorbance values were converted to Percent of Control and plotted against test agent concentrations for EC50 calculations using SoftMax® Pro (version 5.2, Molecular Devices). The plate blank signal average was subtracted from all wells prior to calculating the Percent of Control. Percent of Control values were calculated by dividing the absorbance values for each test well by the No Drug Control average (column 11 values; cells+vehicle control) and multiplying by 100. Plots of Compound Concentration vs. Percent of Control were analyzed using the 4-parameter equation to obtain EC50 values and other parameters that describe the sigmoidal dose response curve.
  • Combination data was analyzed using CompuSyn® software to calculate Combination Index (CI) values to assess synergy. The Fractional Affect (Fa) was calculated from the Percent of Control (from SoftMax® Pro) using the formula: 1−(Percent Control/100). The dosage, fractional affect and molar ratio of compounds tested in combination were entered into the CompuSyn® software for evaluation of the presence/absence of synergy. CompuSyn® assigns a Combination Index (CI) value which rates the level of compounds' effect on proliferation. CI values below 1 indicate the presence of synergy and CI values above 1 indicate antagonism. CI values close to 1 indicate an additive affect. See Chou, PHARMACOL. REV., 58(3):621-81(2006). Table 2 below summarizes the CI values of the synergistic combinations.
  • TABLE 2
    Combination Index Values
    Combination Index
    Combination Cell Line (CI) Values*
    Test drug + cisplatin A549 0.1729
    HCT-116 0.6872
    N87 0.7575
    Test drug + oxaliplatin LoVo 0.219
    Test drug + docetaxel LNCaP 0.5435
    N87 0.6954
    Test drug + 5-fluorouracil HCT-116 0.3608
    LoVo 0.5975
    ZR-75-1 0.6516
    Test drug + gemcitabine A549 0.6472
    PANC-1 0.8952
    Test drug + sorafenib Hep3B2.1-7 0.5361
    A549 0.8469
    Test drug + doxorubicin Hep3B2.1-7 0.252
    Test drug + erlotinib A549 0.5093
    Test drug + everolimus MKL-1 0.354
    *0.1-0.90 = Synergism; 0.90-1.10 = Additive; 1.10-10 = Antagonism.
  • Using CompuSyn software, the combination index (CI) values at different (fa)x (fraction affected) were generated, and the entire spectrum of CIs at different fa values were simulated. The synergism is further illustrated in the isobologram combination index plots in FIGS. 1-15, 31. Note that both the Fa and the CI for the x- and y-axes are dimensionless quantities. Points under the dashed line are synergistic.
  • EXAMPLE 2
  • Cell Culture: The hepatocellular carcinoma cell line Hep3B (from ATCC) was grown in RMPI 1640 supplemented with 10% fetal bovine serum. The epidermal carcinoma-derived cell line KB-3-1 was grown in RPMI 1640+10% FCS. See Shen et al., J. Biol. Chem., 261:7762-7770 (1986).
  • Cytotoxicity Assays: Cells were plated (2×103 cells in 100 μl/well) in 96-well plates and allowed to recover for 24 hours. Drugs were added in another 100 μl growth medium and cells exposed for 72 hours. The proportion of viable cells was determined by MTT assay following the manufacturer's recommendations (EZ4U, Biomedica, Vienna, Austria).
  • As shown in FIGS. 16-22, significant synergies were exhibited by the combination of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] and anti-cancer drugs including erlotinib (FIGS. 16 and 17), BCNU (FIGS. 18 and 19), sunitinib (FIG. 20), and temozolomide (FIGS. 21 and 22) in the cell lines tested.
  • EXAMPLE 3
  • Sorafenib was purchased from LC Laboratories (Woburn, USA). All other substances were purchased from Sigma-Aldrich (St. Louis, USA).
  • Cell Culture: The hepatocellular carcinoma cell line Hep3B was purchased from American Type Culture Collection, Manassas, Va. Cells were grown in RMPI 1640 supplemented with 10% fetal bovine serum. The colon carcinoma cell line HCT116 and respective subline with deleted p53 genes were grown in McCoy's culture medium supplemented with 10% FCS. See Bunz et al, Cancer Res., 62:1129-1133 (2002). Lung cancer cell line A549 was grown in RPMI 1640 medium with 10% FCS, and the hepatocellular carcinoma cell line HepG2 was cultured in the Minimal Essential Medium supplemented with non-essential aminoacids, pyruvate, and 10% FCS. Lung carcinoma cell line VL-8 established in the Institute of Cancer Research, Vienna was grown in RPMI 1640 medium supplemented with 10% FCS. See Berger et al., Int. J. Cancer, 73:84-93 (1997). The mesothelioma cell model P31 and its respective cisplatin-resistant subline P31/cis was grown in Eagle's minimal essential medium with 10% FCS. See Janson et al., Cell Physiol. Biochem., 22:45-56 (2008). Cultures were regularly checked for Mycoplasma contamination.
  • Cytotoxicity Assays: Cells were plated (2×103 cells in 100 μl/well) in 96-well plates and allowed to recover for 24 hours. Drugs were added in another 100 μl growth medium and cells exposed for 72 hours. The proportion of viable cells was determined by MTT assay following the manufacturer's recommendations (EZ4U, Biomedica, Vienna, Austria).
  • As shown in FIGS. 23-29, the combination of sorafenib and sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] gives rise to significant synergies in a variety of cell lines including hepatocellular carcinoma cell line Hep3B, hepatocellular carcinoma cell line HepG2, lung carcinoma cell line VL-8, lung carcinoma cell line A549, mesothelioma cell line P31, colon cancer cell line SW480, and melanoma cell line VM-1.
  • Xenograft Assay: CB17 severe combined immunodeficient (SCID) female mice were used for all in vivo studies. The mice received food and water ad libitum. For tumor application, logarithmically growing Hep3B cells in cell culture were collected by trypsinization and washed once in serum-free culture medium. The cells were then pelleted and resuspended in culture medium to a final cell count of 2×107/ml. 50 μl of the cell suspension was injected s.c. in the right flank of each mouse. Treatment started when all animals in the study had established tumors of a size of about 3×3 mm.
  • Sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] was administered i.v. at a final concentration of 30 mg/kg body weight once a week for 2 weeks (day 1 and day 8). Sorafenib (LC Laboratories, Woburn, Mass., USA) was dissolved in DMSO (50 mg/ml), further diluted in Cremophor EL/95% ethanol (50:50; Sigma), which was followed by a 1:4 dilution in water. 100 μl sorafenib was administered p.o. once daily at 5 consecutive days for two weeks at a dose of 25 mg/kg body weight (days 1-5 and days 8-12).
  • Tumor size was calculated using the equation (l×w2)/2, where l and w refer to the larger and smaller dimensions, respectively, of the tumor. 4 mice were used in each group for each data point.
  • Sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] treatment as a single agent led to a 2.4-fold increase in life span (mean survival 80 days vs. 33 days in control) and thus was superior to sorafenib monotherapy, which induced a 1.9-fold survival increase (60 days). Combination of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] with sorafenib increased the mean survival by 3.9-fold to 96 days. See FIG. 30.
  • EXAMPLE 4
  • The purpose of this experiment was to evaluate the efficacy of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)], administered intravenously (IV) as a single agent and in combination with cisplatin against early stage N87 human gastric carcinoma xenografts in female nude mice.
  • Female athymic mice (Hsd: Athymic Nude-Foxn1nu) were obtained from Harlan. They were 8 weeks old on Day 1 of the experiment. The mice were fed irradiated Rodent Diet 5053 (LabDiet™) and water ad libitum, and grown and experimented on in a clean and controlled environment. Test mice were implanted subcutaneously on Day 0 with 30 to 60 mg tumor fragments. All mice were observed for clinical signs at least once daily. Mice with tumors in excess of 1 g or with ulcerated tumors were euthanized. All procedures carried out in this experiment were conducted in compliance with all the laws, regulations and guidelines of the National Institutes of Health (NIH) and with the necessary approvals.
  • Treatments began on Day 3. All mice weighed ≧18.2 g at the initiation of therapy. Mean group body weights at first treatment were well-matched (range of group means, 22.3-22.8 g). All mice were dosed according to individual body weight on the day of treatment (0.2 ml/20 g). Sixteen days after the initial course of treatment was completed, a second course of treatment was begun for the combination groups only (groups in which cisplatin was dosed at 7.5 mg/kg). A complete second course of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] was given, but only two of the three planned doses of cisplatin were completed due to extensive weight loss.
  • Body weights and tumor measurements were recorded twice weekly. Tumor burden (mg) was estimated from caliper measurements by the formula for the volume of a prolate ellipsoid assuming unit density as: Tumor burden (mg)=(L×W2)/2, where L and W are the respective orthogonal tumor length and width measurements (mm). The primary endpoints used to evaluate efficacy were: % T/C, tumor growth delay, and the number of tumor-free survivors (TFS) at the end of the study. % T/C is defined as the median tumor mass of the Treated Group divided by the median tumor mass of the Control Group×100. In this experiment, % T/C was evaluated when the median Control reached 1 g. Tumor Growth Delay (T−C) was also used to quantify efficacy. Tumor growth delay for this experiment was expressed as a T−C value, where T and C are the median times in days required for the treatment and control group tumors, respectively, to grow to a selected evaluation size, 750 mg.
  • RESULTS: In this experiment, tumor growth delay and Day 28% T/C values (when the medium tumor mass in the Vehicle control Group surpassed 1 g) were used to evaluate the anti-cancer activity of the tested compounds. Treatment with sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (“test drug”) at 30 mg/kg (on days 3, 5, 7, and 27, 29, 31) plus cisplatin at 7.5 mg/kg (on days 3, 7, 11, and 27, 31) produced a significant (P<0.05) tumor growth delay of 16.2 days and a Day 28 T/C value of 16%. The difference in tumor growth delays between the combination regimen and the single agent regimens was significant.
  • EXAMPLE 5
  • The purpose of this experiment was to evaluate the efficacy of sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] as a single agent and in combination with paclitaxel against early stage A549 human lung carcinoma xenografts in female nude mice. Sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] was administered intravenously every two days for three treatments and paclitaxel was administered intravenously for five consecutive days, both beginning on Day 3 post implant. The animals were grown, implanted with tumors and experimented on in the same manner as in Example 5 above, unless otherwise clarified below.
  • Cremophor EL® was used in the context of paclitaxel administration. Specifically, on each day of treatment, the paclitaxel was dissolved in absolute ethanol (10% of the final volume), followed by sequential addition of Cremophor EL® (10% of the final volume) and saline (80% of the final volume) with thorough mixing after each addition.
  • Treatments began on Day 3. All mice weighed ≧17.3 g at the initiation of therapy. Mean group body weights at first treatment were well-matched (range of group means, 20.6-23.5 g). All mice were dosed according to individual body weight on the day of treatment (0.2 ml/20 g).
  • RESULTS: In this experiment, tumor growth delay and Day 38 (the day that the mean tumor burden of the Control group surpassed the evaluation size of 1 g) % T/C values were used to evaluate anti-cancer activity. Treatment with sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] at 30 mg/kg (on days 3, 5, and 7) plus paclitaxel at 20 mg/kg (on days 3, 4, 5, 6, and 7) produced a significant (P<0.05) tumor growth delay of 16.1 days and a Day 38 T/C value of 37% that was also significant.
  • All publications and patent applications mentioned in the specification are indicative of the level of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The mere mentioning of the publications and patent applications does not necessarily constitute an admission that they are prior art to the instant application.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the appended claims.

Claims (20)

What is claimed is:
1. A method of treating cancer, comprising:
administering to a patient in need of treatment, simultaneously or sequentially (1) a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)], and (2) one or more anti-cancer agents chosen from the group consisting of platinum agents, taxane, anthracyclines, 5-FU and prodrugs thereof, nitrosourea compounds, gemcitabine, temozolomide, EGFR inhibitors, mTOR inhibitors, sorafenib, regorafenib, and sunitinib.
2. The method of claim 1, wherein said pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] is sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)].
3. The method of claim 2, wherein said one or more anti-cancer agents includes a platinum agent.
4. The method of claim 3, wherein said platinum agent is cisplatin, carboplatin, or oxaliplatin.
5. The method of claim 2, wherein said one or more anti-cancer agents includes an anthracycline.
6. The method of claim 5, wherein said anthracycline is doxorubicin.
7. The method of claim 2, wherein said one or more anti-cancer agents includes 5-FU or a prodrug thereof.
8. The method of claim 2, wherein said one or more anti-cancer agents includes a nitrosourea compound.
9. The method of claim 8, wherein said nitrosourea compound is BCNU.
10. The method of claim 2, wherein said one or more anti-cancer agents includes gemcitabine.
11. The method of claim 2, wherein said one or more anti-cancer agents includes temozolomide.
12. The method of claim 2, wherein said one or more anti-cancer agents includes an EGFR inhibitor.
13. The method of claim 12, wherein said EGFR inhibitor is erlotinib.
14. The method of claim 2, wherein said one or more anti-cancer agents includes an mTOR inhibitor.
15. The method of claim 14, wherein said mTOR inhibitor is everolimus.
16. The method of claim 2, wherein said one or more anti-cancer agents includes sorafenib or regorafenib.
17. The method of claim 2, wherein said one or more anti-cancer agents includes sunitinib.
18. The method of claim 2, wherein said one or more anti-cancer agents includes a taxane.
19. The method of claim 18, wherein said taxane is docetaxel or paclitaxel.
20. A kit, comprising in a compartmentalized container:
a first unit dosage form of a pharmaceutically acceptable salt of trans-[tetrachlorobis(1H-indazole)ruthenate(III)]; and
a second unit dosage form of one anti-cancer agent chosen from the group consisting of platinum agents, taxane,anthracyclines, 5-FU and prodrugs thereof, nitrosourea compounds, gemcitabine, temozolomide, EGFR inhibitors, mTOR inhibitors, sorafenib, regorafenib, and sunitinib.
US13/744,423 2010-07-18 2013-01-18 Combination therapy using a ruthenium complex Abandoned US20130129840A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/744,423 US20130129840A1 (en) 2010-07-18 2013-01-18 Combination therapy using a ruthenium complex

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36532910P 2010-07-18 2010-07-18
PCT/US2011/044302 WO2012012305A2 (en) 2010-07-18 2011-07-18 Combination therapy using a ruthenium complex
US13/744,423 US20130129840A1 (en) 2010-07-18 2013-01-18 Combination therapy using a ruthenium complex

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/044302 Continuation WO2012012305A2 (en) 2010-07-18 2011-07-18 Combination therapy using a ruthenium complex

Publications (1)

Publication Number Publication Date
US20130129840A1 true US20130129840A1 (en) 2013-05-23

Family

ID=45497392

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/744,423 Abandoned US20130129840A1 (en) 2010-07-18 2013-01-18 Combination therapy using a ruthenium complex

Country Status (7)

Country Link
US (1) US20130129840A1 (en)
EP (1) EP2593101A4 (en)
JP (1) JP6034288B2 (en)
KR (1) KR101783190B1 (en)
CN (1) CN103153303A (en)
AU (1) AU2011279836A1 (en)
WO (1) WO2012012305A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130237510A1 (en) * 2010-10-25 2013-09-12 Niiki Pharma Inc. Method of treating neuroendocrine tumors
US20130281528A1 (en) * 2010-04-19 2013-10-24 Niiki Pharma Acquisition Corp.2 Method of treating gastric cancer
US20170172924A1 (en) * 2013-10-22 2017-06-22 Tyme, Inc. Tyrosine derivatives and compositions comprising them
WO2017151775A1 (en) * 2016-03-01 2017-09-08 Intezyne Technologies Use of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] for the treatment of cancer
WO2018204930A1 (en) * 2017-05-05 2018-11-08 Intezyne Technologies, Inc. Manufacture of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] and compositions thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014001956A2 (en) * 2012-06-25 2014-01-03 Ning Man Cheng Combinational use of pegylated recombinant human arginase with chemotherapeutic/target therapeutic drug in cancer treatment
US20230024119A1 (en) * 2019-12-05 2023-01-26 Bold Therapeutics, Inc. Combined use of sodium trans-[tetrachloridobis(1h-indazole)ruthenate(iii)] and etomoxir for treating cancers
WO2023070199A1 (en) * 2021-10-28 2023-05-04 Bold Therapeutics Inc. Use of sodium trans-[tetrachloridobis(1h-indazole)ruthenate(iii)] to ameliorate proteasome inhibitor resistance

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7338946B2 (en) * 2001-01-26 2008-03-04 Faustus Forschungs Cie. Translational Cancer Research Gmbh Compositions containing a ruthenium(III) complex and a heterocycle
US20090246291A1 (en) * 2008-03-27 2009-10-01 Angelika Burger Method and compositions for treatment of cancer
US20120115833A1 (en) * 2009-04-17 2012-05-10 Niiki Pharma Inc. Method of treating hepatocellular carcinoma

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1230145B (en) * 1989-05-05 1991-10-14 Boehringer Biochemia Srl RUTHENIUM (III) COMPLEXES AS ANTINEOPLASTIC AGENTS.

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7338946B2 (en) * 2001-01-26 2008-03-04 Faustus Forschungs Cie. Translational Cancer Research Gmbh Compositions containing a ruthenium(III) complex and a heterocycle
US20090246291A1 (en) * 2008-03-27 2009-10-01 Angelika Burger Method and compositions for treatment of cancer
US20120115833A1 (en) * 2009-04-17 2012-05-10 Niiki Pharma Inc. Method of treating hepatocellular carcinoma

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Depenbrock et al (Eur. J. Cancer; 33(14); 2404-2410 (1997) *
Heffeter et al. (The Journal of Pharmacology AND Experimental Therapeutic, 312(1); (2005). *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130281528A1 (en) * 2010-04-19 2013-10-24 Niiki Pharma Acquisition Corp.2 Method of treating gastric cancer
US20130237510A1 (en) * 2010-10-25 2013-09-12 Niiki Pharma Inc. Method of treating neuroendocrine tumors
US20170172924A1 (en) * 2013-10-22 2017-06-22 Tyme, Inc. Tyrosine derivatives and compositions comprising them
US11058638B2 (en) * 2013-10-22 2021-07-13 Tyme, Inc. Tyrosine derivatives and compositions comprising them
CN109195600A (en) * 2016-03-01 2019-01-11 茵肽锌科技公司 Trans--[tetrachloro bis- (1H- indazoles) ruthenium (III) hydrochlorate] is used for the purposes for the treatment of cancer
IL261384A (en) * 2016-03-01 2018-10-31 Intezyne Tech Use of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] for the treatment of cancer
US10821095B2 (en) * 2016-03-01 2020-11-03 Bold Therapeutics, Inc. Use of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] for the treatment of cancer
WO2017151775A1 (en) * 2016-03-01 2017-09-08 Intezyne Technologies Use of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] for the treatment of cancer
US11633380B2 (en) 2016-03-01 2023-04-25 Bold Therapeutics, Inc. Use of trans-[tetrachlorobis(1H-indazole)ruthenate(III)] for the treatment of cancer
IL261384B2 (en) * 2016-03-01 2023-05-01 Intezyne Tech Use of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] for the treatment of cancer
WO2018204930A1 (en) * 2017-05-05 2018-11-08 Intezyne Technologies, Inc. Manufacture of trans-[tetrachlorobis(1h-indazole)ruthenate (iii)] and compositions thereof
US10611787B2 (en) 2017-05-05 2020-04-07 Bold Therapeutics, Inc. Manufacture of trans-[tetrachlorobis(1H-indazole)ruthenate (III)] and compositions thereof
US11040993B2 (en) 2017-05-05 2021-06-22 Bold Therapeutics, Inc. Manufacture of trans-[tetrachlorobis(1H-indazole)ruthenate (III)] and compositions thereof
US11739111B2 (en) 2017-05-05 2023-08-29 Bold Therapeutics, Inc. Manufacture of trans-[tetrachlorobis(1H-indazole)ruthenate (III)] and compositions thereof

Also Published As

Publication number Publication date
JP2013531065A (en) 2013-08-01
EP2593101A2 (en) 2013-05-22
KR101783190B1 (en) 2017-09-29
WO2012012305A3 (en) 2012-07-19
CN103153303A (en) 2013-06-12
EP2593101A4 (en) 2014-01-01
KR20130141443A (en) 2013-12-26
JP6034288B2 (en) 2016-11-30
AU2011279836A1 (en) 2013-02-07
WO2012012305A2 (en) 2012-01-26

Similar Documents

Publication Publication Date Title
US20130129840A1 (en) Combination therapy using a ruthenium complex
RU2605335C2 (en) Combination therapy with an antitumor alkaloid
KR101434009B1 (en) Antineoplastic combinations of 4-anilino-3-cyanoquinolines and capecitabine
EP3148536B1 (en) Pharmaceutical combinations for treating cancer
JP2012500180A5 (en)
JP2014132009A5 (en)
US20060293323A1 (en) Therapeutic Combinations of erb B Kinase Inhibitors and Antineoplastic Therapies
US9119877B2 (en) Therapeutic combination comprising a Cdc7 inhibitor and an anti-neoplastic agent
KR20110025178A (en) Combination therapy with an antitumor alkaloid
CN103889418A (en) Compositions and methods for treating cancer using PI3K beta inhibitor and MAPK pathway inhibitor, including MEK and RAF inhibitors
CN111918656B (en) Anticancer pharmaceutical composition for combination therapy
JP7311177B2 (en) Combined use of A-NOR-5α androstane drugs with anticancer drugs
JP7357386B2 (en) Application of the compound or its pharmaceutically acceptable salt, dimer or trimer in the preparation of drugs for cancer treatment
TW201306833A (en) Combination comprising a derivative of the family of the combretastatins and cetuximab
JP2023511072A (en) COMBINATIONS AND THEIR USE FOR THE TREATMENT OF CANCER
WO2023196545A1 (en) Cancer treatments using mta-cooperative prmt5 inhibitors
JP6116482B2 (en) Combination therapy using gallium complex
WO2024031050A2 (en) Hyaluronan-nimesulide conjugate for treating locally advanced or metastatic tumors

Legal Events

Date Code Title Description
AS Assignment

Owner name: NIIKI PHARMA ACQUISITION CORP. 2, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NIIKI PHARMA INC.;REEL/FRAME:031017/0091

Effective date: 20130702

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION